1
|
Avetisyan A, Barria R, Sheehan A, Freeman MR. An Ionic Sensor acts in Parallel to dSarm to Promote Neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620922. [PMID: 39651259 PMCID: PMC11623519 DOI: 10.1101/2024.10.29.620922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
How neurons to sense when they are terminally dysfunctional and activate neurodegeneration remains poorly defined. The pro-degenerative NAD + hydrolase dSarm/SARM1 can act as a metabolic sensor by detecting pathological changes in NAD + /NMN and subsequently induce catastrophic axon degeneration. Here we show Drosophila with-no-lysine kinase (dWnk), which can directly sense Cl - , K + and osmotic pressure, is required for neurodegeneration induced by depletion of the NAD + biosynthetic enzyme dNmnat. dWnk functions in parallel to dSarm and acts through the downstream kinase Frayed to promote axon degeneration and neuronal cell death. dWnk and dSarm ultimately converge on the BTB-Back domain molecule Axundead (Axed) to execute neurodegeneration. Our work argues that neurons use direct sensors of both metabolism (dSarm/SARM1) and ionic/osmotic status (dWnk) to evaluate cellular health and, when dysfunctional, promote neurodegeneration though a common axon death signaling molecule, Axundead.
Collapse
|
2
|
Yarikipati P, Jonusaite S, Pleinis JM, Dominicci Cotto C, Sanchez-Hernandez D, Morrison DE, Goyal S, Schellinger J, Pénalva C, Curtiss J, Rodan AR, Jenny A. Unanticipated domain requirements for Drosophila Wnk kinase in vivo. PLoS Genet 2023; 19:e1010975. [PMID: 37819975 PMCID: PMC10593226 DOI: 10.1371/journal.pgen.1010975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 10/23/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023] Open
Abstract
WNK (With no Lysine [K]) kinases have critical roles in the maintenance of ion homeostasis and the regulation of cell volume. Their overactivation leads to pseudohypoaldosteronism type II (Gordon syndrome) characterized by hyperkalemia and high blood pressure. More recently, WNK family members have been shown to be required for the development of the nervous system in mice, zebrafish, and flies, and the cardiovascular system of mice and fish. Furthermore, human WNK2 and Drosophila Wnk modulate canonical Wnt signaling. In addition to a well-conserved kinase domain, animal WNKs have a large, poorly conserved C-terminal domain whose function has been largely mysterious. In most but not all cases, WNKs bind and activate downstream kinases OSR1/SPAK, which in turn regulate the activity of various ion transporters and channels. Here, we show that Drosophila Wnk regulates Wnt signaling and cell size during the development of the wing in a manner dependent on Fray, the fly homolog of OSR1/SPAK. We show that the only canonical RF(X)V/I motif of Wnk, thought to be essential for WNK interactions with OSR1/SPAK, is required to interact with Fray in vitro. However, this motif is unexpectedly dispensable for Fray-dependent Wnk functions in vivo during fly development and fluid secretion in the Malpighian (renal) tubules. In contrast, a structure function analysis of Wnk revealed that the less-conserved C-terminus of Wnk, that recently has been shown to promote phase transitions in cell culture, is required for viability in vivo. Our data thus provide novel insights into unexpected in vivo roles of specific WNK domains.
Collapse
Affiliation(s)
- Prathibha Yarikipati
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, United States of America
| | - Sima Jonusaite
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
| | - John M. Pleinis
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
| | - Carihann Dominicci Cotto
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, United States of America
| | - David Sanchez-Hernandez
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, United States of America
| | - Daryl E. Morrison
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
| | - Suhani Goyal
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern, Dallas, Texas, United States of America
| | - Jeffrey Schellinger
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern, Dallas, Texas, United States of America
| | - Clothilde Pénalva
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
| | - Jennifer Curtiss
- Department of Cell & Developmental Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Aylin R. Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, United States of America
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah, United States of America
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, United States of America
- Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, United States of America
| | - Andreas Jenny
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, United States of America
- Department of Genetics, Albert Einstein College of Medicine, New York, New York, United States of America
| |
Collapse
|
3
|
Kim LH, Kim JY, Xu YY, Lim MA, Koo BS, Kim JH, Yoon SE, Kim YJ, Choi KW, Chang JW, Hong ST. Tctp, a unique Ing5-binding partner, inhibits the chromatin binding of Enok in Drosophila. Proc Natl Acad Sci U S A 2023; 120:e2218361120. [PMID: 37014852 PMCID: PMC10104566 DOI: 10.1073/pnas.2218361120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/26/2023] [Indexed: 04/05/2023] Open
Abstract
The MOZ/MORF histone acetyltransferase complex is highly conserved in eukaryotes and controls transcription, development, and tumorigenesis. However, little is known about how its chromatin localization is regulated. Inhibitor of growth 5 (ING5) tumor suppressor is a subunit of the MOZ/MORF complex. Nevertheless, the in vivo function of ING5 remains unclear. Here, we report an antagonistic interaction between Drosophila Translationally controlled tumor protein (TCTP) (Tctp) and ING5 (Ing5) required for chromatin localization of the MOZ/MORF (Enok) complex and H3K23 acetylation. Yeast two-hybrid screening using Tctp identified Ing5 as a unique binding partner. In vivo, Ing5 controlled differentiation and down-regulated epidermal growth factor receptor signaling, whereas it is required in the Yorkie (Yki) pathway to determine organ size. Ing5 and Enok mutants promoted tumor-like tissue overgrowth when combined with uncontrolled Yki activity. Tctp depletion rescued the abnormal phenotypes of the Ing5 mutation and increased the nuclear translocation of Ing5 and chromatin binding of Enok. Nonfunctional Enok promoted the nuclear translocation of Ing5 by reducing Tctp, indicating a feedback mechanism between Tctp, Ing5, and Enok to regulate histone acetylation. Therefore, Tctp is essential for H3K23 acetylation by controlling the nuclear translocation of Ing5 and chromatin localization of Enok, providing insights into the roles of human TCTP and ING5-MOZ/MORF in tumorigenesis.
Collapse
Affiliation(s)
- Lee-Hyang Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
| | - Ja-Young Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
| | - Yu-Ying Xu
- Department of Anatomy and Cell Biology, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
| | - Mi Ae Lim
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
| | - Bon Seok Koo
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
| | - Jung Hae Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
| | - Sung-Eun Yoon
- Korea Drosophila Resource Center, Gwangju Institute of Science and Technology, Gwangju61005, Republic of Korea
| | - Young-Joon Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju61005, Republic of Korea
| | - Kwang-Wook Choi
- Department of Biological Sciences, Korea Advanced Institute of Science & Technology, Daejeon34141, Republic of Korea
| | - Jae Won Chang
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
| | - Sung-Tae Hong
- Department of Anatomy and Cell Biology, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon35015, Republic of Korea
| |
Collapse
|
4
|
Neville KE, Finegan TM, Lowe N, Bellomio PM, Na D, Bergstralh DT. The Drosophila mitotic spindle orientation machinery requires activation, not just localization. EMBO Rep 2023; 24:e56074. [PMID: 36629398 PMCID: PMC9986814 DOI: 10.15252/embr.202256074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
The orientation of the mitotic spindle at metaphase determines the placement of the daughter cells. Spindle orientation in animals typically relies on an evolutionarily conserved biological machine comprised of at least four proteins - called Pins, Gαi, Mud, and Dynein in flies - that exerts a pulling force on astral microtubules and reels the spindle into alignment. The canonical model for spindle orientation holds that the direction of pulling is determined by asymmetric placement of this machinery at the cell cortex. In most cell types, this placement is thought to be mediated by Pins, and a substantial body of literature is therefore devoted to identifying polarized cues that govern localized cortical enrichment of Pins. In this study we revisit the canonical model and find that it is incomplete. Spindle orientation in the Drosophila follicular epithelium and embryonic ectoderm requires not only Pins localization but also direct interaction between Pins and the multifunctional protein Discs large. This requirement can be over-ridden by interaction with another Pins interacting protein, Inscuteable.
Collapse
Affiliation(s)
| | - Tara M Finegan
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | - Nicholas Lowe
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | | | - Daxiang Na
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | - Dan T Bergstralh
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
- Department of Physics & AstronomyUniversity of RochesterRochesterNew YorkUSA
- Department of Biomedical GeneticsUniversity of Rochester Medical CenterRochesterNew YorkUSA
| |
Collapse
|
5
|
Kozlov EN, Tokmatcheva EV, Khrustaleva AM, Grebenshchikov ES, Deev RV, Gilmutdinov RA, Lebedeva LA, Zhukova M, Savvateeva-Popova EV, Schedl P, Shidlovskii YV. Long-Term Memory Formation in Drosophila Depends on the 3'UTR of CPEB Gene orb2. Cells 2023; 12:cells12020318. [PMID: 36672258 PMCID: PMC9856895 DOI: 10.3390/cells12020318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/30/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Activation of local translation in neurites in response to stimulation is an important step in the formation of long-term memory (LTM). CPEB proteins are a family of translation factors involved in LTM formation. The Drosophila CPEB protein Orb2 plays an important role in the development and function of the nervous system. Mutations of the coding region of the orb2 gene have previously been shown to impair LTM formation. We found that a deletion of the 3'UTR of the orb2 gene similarly results in loss of LTM in Drosophila. As a result of the deletion, the content of the Orb2 protein remained the same in the neuron soma, but significantly decreased in synapses. Using RNA immunoprecipitation followed by high-throughput sequencing, we detected more than 6000 potential Orb2 mRNA targets expressed in the Drosophila brain. Importantly, deletion of the 3'UTR of orb2 mRNA also affected the localization of the Csp, Pyd, and Eya proteins, which are encoded by putative mRNA targets of Orb2. Therefore, the 3'UTR of the orb2 mRNA is important for the proper localization of Orb2 and other proteins in synapses of neurons and the brain as a whole, providing a molecular basis for LTM formation.
Collapse
Affiliation(s)
- Eugene N. Kozlov
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Elena V. Tokmatcheva
- Institute of Physiology, Russian Academy of Sciences, 188680 St. Petersburg, Russia
| | - Anastasia M. Khrustaleva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Eugene S. Grebenshchikov
- Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), 119992 Moscow, Russia
| | - Roman V. Deev
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Rudolf A. Gilmutdinov
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Lyubov A. Lebedeva
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Mariya Zhukova
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | | | - Paul Schedl
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Department of Molecular Biology, Princeton University, Princeton University, Princeton, NJ 08544-1014, USA
| | - Yulii V. Shidlovskii
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), 119992 Moscow, Russia
- Correspondence:
| |
Collapse
|
6
|
Izadifar A, Courchet J, Virga DM, Verreet T, Hamilton S, Ayaz D, Misbaer A, Vandenbogaerde S, Monteiro L, Petrovic M, Sachse S, Yan B, Erfurth ML, Dascenco D, Kise Y, Yan J, Edwards-Faret G, Lewis T, Polleux F, Schmucker D. Axon morphogenesis and maintenance require an evolutionary conserved safeguard function of Wnk kinases antagonizing Sarm and Axed. Neuron 2021; 109:2864-2883.e8. [PMID: 34384519 DOI: 10.1016/j.neuron.2021.07.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 05/24/2021] [Accepted: 07/08/2021] [Indexed: 12/13/2022]
Abstract
The molecular and cellular mechanisms underlying complex axon morphogenesis are still poorly understood. We report a novel, evolutionary conserved function for the Drosophila Wnk kinase (dWnk) and its mammalian orthologs, WNK1 and 2, in axon branching. We uncover that dWnk, together with the neuroprotective factor Nmnat, antagonizes the axon-destabilizing factors D-Sarm and Axundead (Axed) during axon branch growth, revealing a developmental function for these proteins. Overexpression of D-Sarm or Axed results in axon branching defects, which can be blocked by overexpression of dWnk or Nmnat. Surprisingly, Wnk kinases are also required for axon maintenance of adult Drosophila and mouse cortical pyramidal neurons. Requirement of Wnk for axon maintenance is independent of its developmental function. Inactivation of dWnk or mouse Wnk1/2 in mature neurons leads to axon degeneration in the adult brain. Therefore, Wnk kinases are novel signaling components that provide a safeguard function in both developing and adult axons.
Collapse
Affiliation(s)
- Azadeh Izadifar
- Life and Medical Sciences Institute (LIMES), Bonn, Germany; VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Julien Courchet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, 69622 Villeurbanne, France; Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
| | - Daniel M Virga
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Tine Verreet
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Stevie Hamilton
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Derya Ayaz
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Anke Misbaer
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Sofie Vandenbogaerde
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Laloe Monteiro
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, 69622 Villeurbanne, France
| | - Milan Petrovic
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Sonja Sachse
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Bing Yan
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Maria-Luise Erfurth
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Dan Dascenco
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | | | - Jiekun Yan
- VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Gabriela Edwards-Faret
- Life and Medical Sciences Institute (LIMES), Bonn, Germany; VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Tommy Lewis
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Franck Polleux
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Kavli Institute for Brain Science, Columbia University, New York, NY, USA.
| | - Dietmar Schmucker
- Life and Medical Sciences Institute (LIMES), Bonn, Germany; VIB Center for Brain & Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
7
|
Neural specification, targeting, and circuit formation during visual system assembly. Proc Natl Acad Sci U S A 2021; 118:2101823118. [PMID: 34183440 DOI: 10.1073/pnas.2101823118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Like other sensory systems, the visual system is topographically organized: Its sensory neurons, the photoreceptors, and their targets maintain point-to-point correspondence in physical space, forming a retinotopic map. The iterative wiring of circuits in the visual system conveniently facilitates the study of its development. Over the past few decades, experiments in Drosophila have shed light on the principles that guide the specification and connectivity of visual system neurons. In this review, we describe the main findings unearthed by the study of the Drosophila visual system and compare them with similar events in mammals. We focus on how temporal and spatial patterning generates diverse cell types, how guidance molecules distribute the axons and dendrites of neurons within the correct target regions, how vertebrates and invertebrates generate their retinotopic map, and the molecules and mechanisms required for neuronal migration. We suggest that basic principles used to wire the fly visual system are broadly applicable to other systems and highlight its importance as a model to study nervous system development.
Collapse
|
8
|
Takechi H, Hakeda-Suzuki S, Nitta Y, Ishiwata Y, Iwanaga R, Sato M, Sugie A, Suzuki T. Glial insulin regulates cooperative or antagonistic Golden goal/Flamingo interactions during photoreceptor axon guidance. eLife 2021; 10:66718. [PMID: 33666170 PMCID: PMC7987344 DOI: 10.7554/elife.66718] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/02/2021] [Indexed: 11/29/2022] Open
Abstract
Transmembrane protein Golden goal (Gogo) interacts with atypical cadherin Flamingo (Fmi) to direct R8 photoreceptor axons in the Drosophila visual system. However, the precise mechanisms underlying Gogo regulation during columnar- and layer-specific R8 axon targeting are unknown. Our studies demonstrated that the insulin secreted from surface and cortex glia switches the phosphorylation status of Gogo, thereby regulating its two distinct functions. Non-phosphorylated Gogo mediates the initial recognition of the glial protrusion in the center of the medulla column, whereas phosphorylated Gogo suppresses radial filopodia extension by counteracting Flamingo to maintain a one axon-to-one column ratio. Later, Gogo expression ceases during the midpupal stage, thus allowing R8 filopodia to extend vertically into the M3 layer. These results demonstrate that the long- and short-range signaling between the glia and R8 axon growth cones regulates growth cone dynamics in a stepwise manner, and thus shapes the entire organization of the visual system.
Collapse
Affiliation(s)
- Hiroki Takechi
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Satoko Hakeda-Suzuki
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yohei Nitta
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan.,Brain Research Institute, Niigata University, Niigata, Japan
| | - Yuichi Ishiwata
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Riku Iwanaga
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Makoto Sato
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan.,Laboratory of Developmental Neurobiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Atsushi Sugie
- Center for Transdisciplinary Research, Niigata University, Niigata, Japan.,Brain Research Institute, Niigata University, Niigata, Japan
| | - Takashi Suzuki
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
9
|
Kawamura H, Hakeda-Suzuki S, Suzuki T. Activity-dependent endocytosis of Wingless regulates synaptic plasticity in the Drosophila visual system. Genes Genet Syst 2021; 95:235-247. [PMID: 33298662 DOI: 10.1266/ggs.20-00030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Neural activity contributes to synaptic regulation in sensory systems, which allows organisms to adjust to changing environments. However, little is known about how synaptic molecular components are regulated to achieve activity-dependent plasticity at central synapses. Previous studies have shown that following prolonged exposure to natural ambient light, the presynaptic active zone (AZ), an area associated with presynaptic neurotransmitter release in Drosophila photoreceptors, undergoes reversible remodeling. Other studies suggest that the secretory protein Wingless (Wg; an ortholog of Wnt-1) can mediate communication between synaptic cells to achieve synaptic remodeling. However, the source of Wg and the mechanism of Wg signal modulation by neuronal activity remained unclear. Here, we found that Wg secreted from glial cells regulates synaptic remodeling in photoreceptors. In addition, antibody staining revealed that Wg changes its localization depending on light conditions. Although Wg is secreted from glial cells, Wg appeared inside photoreceptor axons when flies were kept under light conditions, suggesting that an increase in neuronal activity causes Wg internalization into photoreceptors by endocytosis. Indeed, by blocking endocytosis in photoreceptors, the localization of Wg in photoreceptors disappeared. Interestingly, Wg accumulation was higher in axons with disassembled AZ structure than in axons whose AZ structure was stabilized at the single-cell level, indicating that Wg endocytosis may trigger AZ disassembly. Furthermore, when we genetically activated Wg signaling, Wg accumulation in photoreceptors decreased. Conversely, when we suppressed Wg signaling there was an increase in Wg accumulation. Through RNAi screening of Ca2+-binding proteins in photoreceptors, we found that Calcineurin is a key molecule that triggers Wg endocytosis. Overall, we propose that Wg signaling is regulated by a negative feedback loop driven by Wg endocytosis. The increase in neuronal activity is transmitted via calcium signaling, which leads to a decrease in Wg signaling and thereby promotes presynaptic remodeling.
Collapse
Affiliation(s)
- Hinata Kawamura
- Graduate School of Life Science and Technology, Tokyo Institute of Technology
| | | | - Takashi Suzuki
- Graduate School of Life Science and Technology, Tokyo Institute of Technology
| |
Collapse
|
10
|
DeAngelis MW, Coolon JD, Johnson RI. Comparative transcriptome analyses of the Drosophila pupal eye. G3-GENES GENOMES GENETICS 2021; 11:5995320. [PMID: 33561221 PMCID: PMC8043229 DOI: 10.1093/g3journal/jkaa003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/08/2020] [Indexed: 12/04/2022]
Abstract
Tissue function is dependent on correct cellular organization and behavior. As a result, the identification and study of genes that contribute to tissue morphogenesis is of paramount importance to the fields of cell and developmental biology. Many of the genes required for tissue patterning and organization are highly conserved between phyla. This has led to the emergence of several model organisms and developmental systems that are used to study tissue morphogenesis. One such model is the Drosophila melanogaster pupal eye that has a highly stereotyped arrangement of cells. In addition, the pupal eye is postmitotic that allows for the study of tissue morphogenesis independent from any effects of proliferation. While the changes in cell morphology and organization that occur throughout pupal eye development are well documented, less is known about the corresponding transcriptional changes that choreograph these processes. To identify these transcriptional changes, we dissected wild-type Canton S pupal eyes and performed RNA-sequencing. Our analyses identified differential expression of many loci that are documented regulators of pupal eye morphogenesis and contribute to multiple biological processes including signaling, axon projection, adhesion, and cell survival. We also identified differential expression of genes not previously implicated in pupal eye morphogenesis such as components of the Toll pathway, several non-classical cadherins, and components of the muscle sarcomere, which could suggest these loci function as novel patterning factors.
Collapse
Affiliation(s)
- Miles W DeAngelis
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| | - Joseph D Coolon
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| | - Ruth I Johnson
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| |
Collapse
|
11
|
Xu W, Xie XJ, Faust AK, Liu M, Li X, Chen F, Naquin AA, Walton AC, Kishbaugh PW, Ji JY. All-Atomic Molecular Dynamic Studies of Human and Drosophila CDK8: Insights into Their Kinase Domains, the LXXLL Motifs, and Drug Binding Site. Int J Mol Sci 2020; 21:E7511. [PMID: 33053834 PMCID: PMC7590003 DOI: 10.3390/ijms21207511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022] Open
Abstract
Cyclin-dependent kinase 8 (CDK8) and its regulatory partner Cyclin C (CycC) play conserved roles in modulating RNA polymerase II (Pol II)-dependent gene expression. To understand the structure and function relations of CDK8, we analyzed the structures of human and Drosophila CDK8 proteins using molecular dynamics simulations, combined with functional analyses in Drosophila. Specifically, we evaluated the structural differences between hCDK8 and dCDK8 to predict the effects of the LXXLL motif mutation (AQKAA), the P154L mutations, and drug binding on local structures of the CDK8 proteins. First, we have observed that both the LXXLL motif and the kinase activity of CDK8 are required for the normal larval-to-pupal transition in Drosophila. Second, our molecular dynamic analyses have revealed that hCDK8 has higher hydrogen bond occupation of His149-Asp151 and Asp151-Asn156 than dCDK8. Third, the substructure of Asp282, Phe283, Arg285, Thr287 and Cys291 can distinguish human and Drosophila CDK8 structures. In addition, there are two hydrogen bonds in the LXXLL motif: a lower occupation between L312 and L315, and a relatively higher occupation between L312 and L316. Human CDK8 has higher hydrogen bond occupation between L312 and L316 than dCDK8. Moreover, L312, L315 and L316 in the LXXLL motif of CDK8 have the specific pattern of hydrogen bonds and geometries, which could be crucial for the binding to nuclear receptors. Furthermore, the P154L mutation dramatically decreases the hydrogen bond between L312 and L315 in hCDK8, but not in dCDK8. The mutations of P154L and AQKAA modestly alter the local structures around residues 154. Finally, we identified the inhibitor-induced conformational changes of hCDK8, and our results suggest a structural difference in the drug-binding site between hCDK8 and dCDK8. Taken together, these results provide the structural insights into the roles of the LXXLL motif and the kinase activity of CDK8 in vivo.
Collapse
Affiliation(s)
- Wu Xu
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA; (A.K.F.); (A.A.N.); (A.C.W.); (P.W.K.)
| | - Xiao-Jun Xie
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, 8447 Riverside Parkway, Bryan, TX 77807, USA; (X.-J.X.); (M.L.); (X.L.)
| | - Ali K. Faust
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA; (A.K.F.); (A.A.N.); (A.C.W.); (P.W.K.)
| | - Mengmeng Liu
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, 8447 Riverside Parkway, Bryan, TX 77807, USA; (X.-J.X.); (M.L.); (X.L.)
| | - Xiao Li
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, 8447 Riverside Parkway, Bryan, TX 77807, USA; (X.-J.X.); (M.L.); (X.L.)
| | - Feng Chen
- High Performance Computing, 329 Frey Computing Services Center, Louisiana State University, Baton Rouge, LA 70803, USA;
| | - Ashlin A. Naquin
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA; (A.K.F.); (A.A.N.); (A.C.W.); (P.W.K.)
| | - Avery C. Walton
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA; (A.K.F.); (A.A.N.); (A.C.W.); (P.W.K.)
| | - Peter W. Kishbaugh
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA; (A.K.F.); (A.A.N.); (A.C.W.); (P.W.K.)
| | - Jun-Yuan Ji
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, 8447 Riverside Parkway, Bryan, TX 77807, USA; (X.-J.X.); (M.L.); (X.L.)
| |
Collapse
|
12
|
Stenesen D, Moehlman AT, Schellinger JN, Rodan AR, Krämer H. The glial sodium-potassium-2-chloride cotransporter is required for synaptic transmission in the Drosophila visual system. Sci Rep 2019; 9:2475. [PMID: 30792494 PMCID: PMC6385505 DOI: 10.1038/s41598-019-38850-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 01/08/2019] [Indexed: 02/07/2023] Open
Abstract
The Drosophila Ncc69 gene encodes a Na+-K+-2Cl−-cotransporter (NKCC) that is critical for regulating intra- and extracellular ionic conditions in different tissues. Here, we show that the Ncc69 transporter is necessary for fly vision and that its expression is required non-autonomously in glia to maintain visual synaptic transmission. Flies mutant for Ncc69 exhibit normal photoreceptor depolarization in response to a light pulse but lack the ON and OFF-transients characteristic of postsynaptic responses of lamina neurons, indicating a failure in synaptic transmission. We also find that synaptic transmission requires the Ncc69 regulatory kinases WNK and Fray in glia. The ERG phenotype is associated with a defect in the recycling of the histamine neurotransmitter. Ncc69 mutants exhibit higher levels of the transport metabolite carcinine in lamina cartridges, with its accumulation most intense in the extracellular space. Our work reveals a novel role of glial NKCC transporters in synaptic transmission, possibly through regulating extracellular ionic conditions.
Collapse
Affiliation(s)
- Drew Stenesen
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.,Biology Department, University of Dallas, Irving, TX, 75062, USA
| | - Andrew T Moehlman
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jeffrey N Schellinger
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Aylin R Rodan
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA. .,Department of Internal Medicine, Division of Nephrology and Hypertension and Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112, USA. .,Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA.
| | - Helmut Krämer
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA. .,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
13
|
Deal SL, Yamamoto S. Unraveling Novel Mechanisms of Neurodegeneration Through a Large-Scale Forward Genetic Screen in Drosophila. Front Genet 2019; 9:700. [PMID: 30693015 PMCID: PMC6339878 DOI: 10.3389/fgene.2018.00700] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 12/13/2018] [Indexed: 01/04/2023] Open
Abstract
Neurodegeneration is characterized by progressive loss of neurons. Genetic and environmental factors both contribute to demise of neurons, leading to diverse devastating cognitive and motor disorders, including Alzheimer's and Parkinson's diseases in humans. Over the past few decades, the fruit fly, Drosophila melanogaster, has become an integral tool to understand the molecular, cellular and genetic mechanisms underlying neurodegeneration. Extensive tools and sophisticated technologies allow Drosophila geneticists to identify and study evolutionarily conserved genes that are essential for neural maintenance. In this review, we will focus on a large-scale mosaic forward genetic screen on the fly X-chromosome that led to the identification of a number of essential genes that exhibit neurodegenerative phenotypes when mutated. Most genes identified from this screen are evolutionarily conserved and many have been linked to human diseases with neurological presentations. Systematic electrophysiological and ultrastructural characterization of mutant tissue in the context of the Drosophila visual system, followed by a series of experiments to understand the mechanism of neurodegeneration in each mutant led to the discovery of novel molecular pathways that are required for neuronal integrity. Defects in mitochondrial function, lipid and iron metabolism, protein trafficking and autophagy are recurrent themes, suggesting that insults that eventually lead to neurodegeneration may converge on a set of evolutionarily conserved cellular processes. Insights from these studies have contributed to our understanding of known neurodegenerative diseases such as Leigh syndrome and Friedreich's ataxia and have also led to the identification of new human diseases. By discovering new genes required for neural maintenance in flies and working with clinicians to identify patients with deleterious variants in the orthologous human genes, Drosophila biologists can play an active role in personalized medicine.
Collapse
Affiliation(s)
- Samantha L Deal
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, United States
| | - Shinya Yamamoto
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
14
|
Stratmann J, Ekman H, Thor S. Branching gene regulatory network dictating different aspects of a neuronal cell identity. Development 2019; 146:dev.174300. [DOI: 10.1242/dev.174300] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/22/2019] [Indexed: 12/30/2022]
Abstract
The nervous system displays a daunting cellular diversity. Neuronal sub-types differ from each other in several aspects, including their neurotransmitter expression and axon projection. These aspects can converge, but can also diverge, such that neurons expressing the same neurotransmitter may project axons to different targets. It is not well understood how regulatory programs converge/diverge to associate/dissociate different cell fate features. Studies of the Drosophila Tv1 neurons have identified a regulatory cascade; ladybird early -> collier -> apterous/eyes absent -> dimmed, which specifies Tv1 neurotransmitter expression. Here, we conduct genetic and transcriptome analysis to address how other aspects of Tv1 cell fate is governed. We find that an initiator terminal selector gene triggers a feedforward loop which branches into different subroutines, each of which establishes different features of this one unique neuronal cell fate.
Collapse
Affiliation(s)
- Johannes Stratmann
- Department of Clinical and Experimental Medicine, Linkoping University, SE-581 85 Linkoping, Sweden
| | - Helen Ekman
- Department of Clinical and Experimental Medicine, Linkoping University, SE-581 85 Linkoping, Sweden
| | - Stefan Thor
- Department of Clinical and Experimental Medicine, Linkoping University, SE-581 85 Linkoping, Sweden
- School of Biomedical Sciences, University of Queensland, St Lucia QLD 4072, Australia
| |
Collapse
|
15
|
Fochler S, Morozova TV, Davis MR, Gearhart AW, Huang W, Mackay TFC, Anholt RRH. Genetics of alcohol consumption in Drosophila melanogaster. GENES, BRAIN, AND BEHAVIOR 2017; 16:675-685. [PMID: 28627812 PMCID: PMC5667673 DOI: 10.1111/gbb.12399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/24/2017] [Accepted: 06/14/2017] [Indexed: 12/30/2022]
Abstract
Individual variation in alcohol consumption in human populations is determined by genetic, environmental, social and cultural factors. In contrast to humans, genetic contributions to complex behavioral phenotypes can be readily dissected in Drosophila, where both the genetic background and environment can be controlled and behaviors quantified through simple high-throughput assays. Here, we measured voluntary consumption of ethanol in ∼3000 individuals of each sex from an advanced intercross population derived from 37 lines of the Drosophila melanogaster Genetic Reference Panel. Extreme quantitative trait loci mapping identified 385 differentially segregating allelic variants located in or near 291 genes at P < 10-8 . The effects of single nucleotide polymorphisms associated with voluntary ethanol consumption are sex-specific, as found for other alcohol-related phenotypes. To assess causality, we used RNA interference knockdown or P{MiET1} mutants and their corresponding controls and functionally validated 86% of candidate genes in at least one sex. We constructed a genetic network comprised of 23 genes along with a separate trio and a pair of connected genes. Gene ontology analyses showed enrichment of developmental genes, including development of the nervous system. Furthermore, a network of human orthologs showed enrichment for signal transduction processes, protein metabolism and developmental processes, including nervous system development. Our results show that the genetic architecture that underlies variation in voluntary ethanol consumption is sexually dimorphic and partially overlaps with genetic factors that control variation in feeding behavior and alcohol sensitivity. This integrative genetic architecture is rooted in evolutionarily conserved features that can be extrapolated to human genetic interaction networks.
Collapse
Affiliation(s)
- S Fochler
- W. M. Keck Center for Behavioral Biology, Program in Genetics, and Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - T V Morozova
- W. M. Keck Center for Behavioral Biology, Program in Genetics, and Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - M R Davis
- W. M. Keck Center for Behavioral Biology, Program in Genetics, and Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - A W Gearhart
- W. M. Keck Center for Behavioral Biology, Program in Genetics, and Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - W Huang
- W. M. Keck Center for Behavioral Biology, Program in Genetics, and Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - T F C Mackay
- W. M. Keck Center for Behavioral Biology, Program in Genetics, and Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - R R H Anholt
- W. M. Keck Center for Behavioral Biology, Program in Genetics, and Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
16
|
DISCO interacting protein 2 determines direction of axon projection under the regulation of c-Jun N-terminal kinase in the Drosophila mushroom body. Biochem Biophys Res Commun 2017; 487:116-121. [DOI: 10.1016/j.bbrc.2017.04.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 04/07/2017] [Indexed: 11/21/2022]
|
17
|
Wang JB, Lu HL, St. Leger RJ. The genetic basis for variation in resistance to infection in the Drosophila melanogaster genetic reference panel. PLoS Pathog 2017; 13:e1006260. [PMID: 28257468 PMCID: PMC5352145 DOI: 10.1371/journal.ppat.1006260] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/15/2017] [Accepted: 02/24/2017] [Indexed: 01/01/2023] Open
Abstract
Individuals vary extensively in the way they respond to disease but the genetic basis of this variation is not fully understood. We found substantial individual variation in resistance and tolerance to the fungal pathogen Metarhizium anisopliae Ma549 using the Drosophila melanogaster Genetic Reference Panel (DGRP). In addition, we found that host defense to Ma549 was correlated with defense to the bacterium Pseudomonas aeruginosa Pa14, and several previously published DGRP phenotypes including oxidative stress sensitivity, starvation stress resistance, hemolymph glucose levels, and sleep indices. We identified polymorphisms associated with differences between lines in both their mean survival times and microenvironmental plasticity, suggesting that lines differ in their ability to adapt to variable pathogen exposures. The majority of polymorphisms increasing resistance to Ma549 were sex biased, located in non-coding regions, had moderately large effect and were rare, suggesting that there is a general cost to defense. Nevertheless, host defense was not negatively correlated with overall longevity and fecundity. In contrast to Ma549, minor alleles were concentrated in the most Pa14-susceptible as well as the most Pa14-resistant lines. A pathway based analysis revealed a network of Pa14 and Ma549-resistance genes that are functionally connected through processes that encompass phagocytosis and engulfment, cell mobility, intermediary metabolism, protein phosphorylation, axon guidance, response to DNA damage, and drug metabolism. Functional testing with insertional mutagenesis lines indicates that 12/13 candidate genes tested influence susceptibility to Ma549. Many candidate genes have homologs identified in studies of human disease, suggesting that genes affecting variation in susceptibility are conserved across species.
Collapse
Affiliation(s)
- Jonathan B. Wang
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
| | - Hsiao-Ling Lu
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
| | - Raymond J. St. Leger
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
18
|
Mutations in the Chromatin Regulator Gene BRPF1 Cause Syndromic Intellectual Disability and Deficient Histone Acetylation. Am J Hum Genet 2017; 100:91-104. [PMID: 27939640 DOI: 10.1016/j.ajhg.2016.11.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/10/2016] [Indexed: 12/18/2022] Open
Abstract
Identification of over 500 epigenetic regulators in humans raises an interesting question regarding how chromatin dysregulation contributes to different diseases. Bromodomain and PHD finger-containing protein 1 (BRPF1) is a multivalent chromatin regulator possessing three histone-binding domains, one non-specific DNA-binding module, and several motifs for interacting with and activating three lysine acetyltransferases. Genetic analyses of fish brpf1 and mouse Brpf1 have uncovered an important role in skeletal, hematopoietic, and brain development, but it remains unclear how BRPF1 is linked to human development and disease. Here, we describe an intellectual disability disorder in ten individuals with inherited or de novo monoallelic BRPF1 mutations. Symptoms include infantile hypotonia, global developmental delay, intellectual disability, expressive language impairment, and facial dysmorphisms. Central nervous system and spinal abnormalities are also seen in some individuals. These clinical features overlap with but are not identical to those reported for persons with KAT6A or KAT6B mutations, suggesting that BRPF1 targets these two acetyltransferases and additional partners in humans. Functional assays showed that the resulting BRPF1 variants are pathogenic and impair acetylation of histone H3 at lysine 23, an abundant but poorly characterized epigenetic mark. We also found a similar deficiency in different lines of Brpf1-knockout mice. These data indicate that aberrations in the chromatin regulator gene BRPF1 cause histone H3 acetylation deficiency and a previously unrecognized intellectual disability syndrome.
Collapse
|
19
|
Abstract
WNK (With-No-Lysine (K)) kinases are serine-threonine kinases characterized by an atypical placement of a catalytic lysine within the kinase domain. Mutations in human WNK1 or WNK4 cause an autosomal dominant syndrome of hypertension and hyperkalemia, reflecting the fact that WNK kinases are critical regulators of renal ion transport processes. Here, the role of WNKs in the regulation of ion transport processes in vertebrate and invertebrate renal function, cellular and organismal osmoregulation, and cell migration and cerebral edema will be reviewed, along with emerging literature demonstrating roles for WNKs in cardiovascular and neural development, Wnt signaling, and cancer. Conserved roles for these kinases across phyla are emphasized.
Collapse
Affiliation(s)
| | - Andreas Jenny
- Albert Einstein College of Medicine, New York, NY, United States.
| |
Collapse
|
20
|
Transcriptome Profiling Identifies Multiplexin as a Target of SAGA Deubiquitinase Activity in Glia Required for Precise Axon Guidance During Drosophila Visual Development. G3-GENES GENOMES GENETICS 2016; 6:2435-45. [PMID: 27261002 PMCID: PMC4978897 DOI: 10.1534/g3.116.031310] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Spt-Ada-Gcn5 Acetyltransferase (SAGA) complex is a transcriptional coactivator with histone acetylase and deubiquitinase activities that plays an important role in visual development and function. In Drosophila melanogaster, four SAGA subunits are required for the deubiquitination of monoubiquitinated histone H2B (ubH2B): Nonstop, Sgf11, E(y)2, and Ataxin 7. Mutations that disrupt SAGA deubiquitinase activity cause defects in neuronal connectivity in the developing Drosophila visual system. In addition, mutations in SAGA result in the human progressive visual disorder spinocerebellar ataxia type 7 (SCA7). Glial cells play a crucial role in both the neuronal connectivity defect in nonstop and sgf11 flies, and in the retinal degeneration observed in SCA7 patients. Thus, we sought to identify the gene targets of SAGA deubiquitinase activity in glia in the Drosophila larval central nervous system. To do this, we enriched glia from wild-type, nonstop, and sgf11 larval optic lobes using affinity-purification of KASH-GFP tagged nuclei, and then examined each transcriptome using RNA-seq. Our analysis showed that SAGA deubiquitinase activity is required for proper expression of 16% of actively transcribed genes in glia, especially genes involved in proteasome function, protein folding and axon guidance. We further show that the SAGA deubiquitinase-activated gene Multiplexin (Mp) is required in glia for proper photoreceptor axon targeting. Mutations in the human ortholog of Mp, COL18A1, have been identified in a family with a SCA7-like progressive visual disorder, suggesting that defects in the expression of this gene in SCA7 patients could play a role in the retinal degeneration that is unique to this ataxia.
Collapse
|
21
|
Regulation of KAT6 Acetyltransferases and Their Roles in Cell Cycle Progression, Stem Cell Maintenance, and Human Disease. Mol Cell Biol 2016; 36:1900-7. [PMID: 27185879 DOI: 10.1128/mcb.00055-16] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The lysine acetyltransferase 6 (KAT6) histone acetyltransferase (HAT) complexes are highly conserved from yeast to higher organisms. They acetylate histone H3 and other nonhistone substrates and are involved in cell cycle regulation and stem cell maintenance. In addition, the human KAT6 HATs are recurrently mutated in leukemia and solid tumors. Therefore, it is important to understand the mechanisms underlying the regulation of KAT6 HATs and their roles in cell cycle progression. In this minireview, we summarize the identification and analysis of the KAT6 complexes and discuss the regulatory mechanisms governing their enzymatic activities and substrate specificities. We further focus on the roles of KAT6 HATs in regulating cell proliferation and stem cell maintenance and review recent insights that aid in understanding their involvement in human diseases.
Collapse
|
22
|
Dascenco D, Erfurth ML, Izadifar A, Song M, Sachse S, Bortnick R, Urwyler O, Petrovic M, Ayaz D, He H, Kise Y, Thomas F, Kidd T, Schmucker D. Slit and Receptor Tyrosine Phosphatase 69D Confer Spatial Specificity to Axon Branching via Dscam1. Cell 2015; 162:1140-54. [PMID: 26317474 PMCID: PMC4699798 DOI: 10.1016/j.cell.2015.08.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 06/30/2015] [Accepted: 07/10/2015] [Indexed: 11/26/2022]
Abstract
Axonal branching contributes substantially to neuronal circuit complexity. Studies in Drosophila have shown that loss of Dscam1 receptor diversity can fully block axon branching in mechanosensory neurons. Here we report that cell-autonomous loss of the receptor tyrosine phosphatase 69D (RPTP69D) and loss of midline-localized Slit inhibit formation of specific axon collaterals through modulation of Dscam1 activity. Genetic and biochemical data support a model in which direct binding of Slit to Dscam1 enhances the interaction of Dscam1 with RPTP69D, stimulating Dscam1 dephosphorylation. Single-growth-cone imaging reveals that Slit/RPTP69D are not required for general branch initiation but instead promote the extension of specific axon collaterals. Hence, although regulation of intrinsic Dscam1-Dscam1 isoform interactions is essential for formation of all mechanosensory-axon branches, the local ligand-induced alterations of Dscam1 phosphorylation in distinct growth-cone compartments enable the spatial specificity of axon collateral formation.
Collapse
Affiliation(s)
- Dan Dascenco
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Maria-Luise Erfurth
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium; Institute of Biochemistry, Christian-Albrechts-University of Kiel, Olshausenstr. 40, 24098 Kiel, Germany
| | - Azadeh Izadifar
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Minmin Song
- Biology/MS 314, University of Nevada, Reno, NV 89557, USA
| | - Sonja Sachse
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium; Department of Biology, Chemistry & Pharmacy, Free University Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Rachel Bortnick
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Olivier Urwyler
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Milan Petrovic
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Derya Ayaz
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Haihuai He
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium
| | - Yoshiaki Kise
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Franziska Thomas
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Thomas Kidd
- Biology/MS 314, University of Nevada, Reno, NV 89557, USA
| | - Dietmar Schmucker
- Neuronal Wiring Laboratory, VIB, Herestraat 49, 3000 Leuven, Belgium; Department of Oncology, School of Medicine, University of Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
23
|
Garcia JD, Dewey EB, Johnston CA. Dishevelled binds the Discs large 'Hook' domain to activate GukHolder-dependent spindle positioning in Drosophila. PLoS One 2014; 9:e114235. [PMID: 25461409 PMCID: PMC4252473 DOI: 10.1371/journal.pone.0114235] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/05/2014] [Indexed: 11/19/2022] Open
Abstract
Communication between cortical cell polarity cues and the mitotic spindle ensures proper orientation of cell divisions within complex tissues. Defects in mitotic spindle positioning have been linked to various developmental disorders and have recently emerged as a potential contributor to tumorigenesis. Despite the importance of this process to human health, the molecular mechanisms that regulate spindle orientation are not fully understood. Moreover, it remains unclear how diverse cortical polarity complexes might cooperate to influence spindle positioning. We and others have demonstrated spindle orientation roles for Dishevelled (Dsh), a key regulator of planar cell polarity, and Discs large (Dlg), a conserved apico-basal cell polarity regulator, effects which were previously thought to operate within distinct molecular pathways. Here we identify a novel direct interaction between the Dsh-PDZ domain and the alternatively spliced “I3-insert” of the Dlg-Hook domain, thus establishing a potential convergent Dsh/Dlg pathway. Furthermore, we identify a Dlg sequence motif necessary for the Dsh interaction that shares homology to the site of Dsh binding in the Frizzled receptor. Expression of Dsh enhanced Dlg-mediated spindle positioning similar to deletion of the Hook domain. This Dsh-mediated activation was dependent on the Dlg-binding partner, GukHolder (GukH). These results suggest that Dsh binding may regulate core interdomain conformational dynamics previously described for Dlg. Together, our results identify Dlg as an effector of Dsh signaling and demonstrate a Dsh-mediated mechanism for the activation of Dlg/GukH-dependent spindle positioning. Cooperation between these two evolutionarily-conserved cell polarity pathways could have important implications to both the development and maintenance of tissue homeostasis in animals.
Collapse
Affiliation(s)
- Joshua D. Garcia
- Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Evan B. Dewey
- Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Christopher A. Johnston
- Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
- * E-mail:
| |
Collapse
|
24
|
Li KL, Lu TM, Yu JK. Genome-wide survey and expression analysis of the bHLH-PAS genes in the amphioxus Branchiostoma floridae reveal both conserved and diverged expression patterns between cephalochordates and vertebrates. EvoDevo 2014; 5:20. [PMID: 24959341 PMCID: PMC4066832 DOI: 10.1186/2041-9139-5-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 05/07/2014] [Indexed: 01/13/2023] Open
Abstract
Background The bHLH-PAS transcription factors are found in both protostomes and deuterostomes. They are involved in many developmental and physiological processes, including regional differentiation of the central nervous system, tube-formation, hypoxia signaling, aromatic hydrocarbon sensing, and circadian rhythm regulation. To understand the evolution of these genes in chordates, we analyzed the bHLH-PAS genes of the basal chordate amphioxus (Branchiostoma floridae). Results From the amphioxus draft genome database, we identified ten bHLH-PAS genes, nine of which could be assigned to known orthologous families. The tenth bHLH-PAS gene could not be assigned confidently to any known bHLH family; however, phylogenetic analysis clustered this gene with arthropod Met family genes and two spiralian bHLH-PAS-containing sequences, suggesting that they may share the same ancestry. We examined temporal and spatial expression patterns of these bHLH-PAS genes in developing amphioxus embryos. We found that BfArnt, BfNcoa, BfSim, and BfHifα were expressed in the central nervous system in patterns similar to those of their vertebrate homologs, suggesting that their functions may be conserved. By contrast, the amphioxus BfAhr and BfNpas4 had expression patterns distinct from those in vertebrates. These results imply that there were changes in gene regulation after the divergence of cephalochordates and vertebrates. Conclusions We have identified ten bHLH-PAS genes from the amphioxus genome and determined the embryonic expression profiles for these genes. In addition to the nine currently recognized bHLH-PAS families, our survey suggests that the BfbHLHPAS-orphan gene along with arthropod Met genes and the newly identified spiralian bHLH-PAS-containing sequences represent an ancient group of genes that were lost in the vertebrate lineage. In a comparison with the expression patterns of the vertebrate bHLH-PAS paralogs, which are the result of whole-genome duplication, we found that although several members seem to retain conserved expression patterns during chordate evolution, many duplicated paralogs may have undergone subfunctionalization and neofunctionalization in the vertebrate lineage. In addition, our survey of amphioxus bHLH-PAS gene models from genome browser with experimentally verified cDNA sequences calls into question the accuracy of the current in silico gene annotation of the B. floridae genome.
Collapse
Affiliation(s)
- Kun-Lung Li
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan ; Institute of Oceanography, National Taiwan University, Taipei 10617, Taiwan
| | - Tsai-Ming Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Jr-Kai Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan ; Institute of Oceanography, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
25
|
Bulat V, Rast M, Pielage J. Presynaptic CK2 promotes synapse organization and stability by targeting Ankyrin2. ACTA ACUST UNITED AC 2014; 204:77-94. [PMID: 24395637 PMCID: PMC3882785 DOI: 10.1083/jcb.201305134] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Phosphorylation of synaptic cytoskeletal components by casein kinase 2 promotes the development and maintenance of synaptic connections. The precise regulation of synapse maintenance is critical to the development and function of neuronal circuits. Using an in vivo RNAi screen targeting the Drosophila kinome and phosphatome, we identify 11 kinases and phosphatases controlling synapse stability by regulating cytoskeletal, phospholipid, or metabolic signaling. We focus on casein kinase 2 (CK2) and demonstrate that the regulatory (β) and catalytic (α) subunits of CK2 are essential for synapse maintenance. CK2α kinase activity is required in the presynaptic motoneuron, and its interaction with CK2β, mediated cooperatively by two N-terminal residues of CK2α, is essential for CK2 holoenzyme complex stability and function in vivo. Using genetic and biochemical approaches we identify Ankyrin2 as a key presynaptic target of CK2 to maintain synapse stability. In addition, CK2 activity controls the subcellular organization of individual synaptic release sites within the presynaptic nerve terminal. Our study identifies phosphorylation of structural synaptic components as a compelling mechanism to actively control the development and longevity of synaptic connections.
Collapse
Affiliation(s)
- Victoria Bulat
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | | | | |
Collapse
|
26
|
Xin T, Xuan T, Tan J, Li M, Zhao G, Li M. The Drosophila putative histone acetyltransferase Enok maintains female germline stem cells through regulating Bruno and the niche. Dev Biol 2013; 384:1-12. [PMID: 24120347 DOI: 10.1016/j.ydbio.2013.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/29/2013] [Accepted: 10/02/2013] [Indexed: 12/16/2022]
Abstract
Maintenance of adult stem cells is largely dependent on the balance between their self-renewal and differentiation. The Drosophila ovarian germline stem cells (GSCs) provide a powerful in vivo system for studying stem cell fate regulation. It has been shown that maintaining the GSC population involves both genetic and epigenetic mechanisms. Although the role of epigenetic regulation in this process is evident, the underlying mechanisms remain to be further explored. In this study, we find that Enoki mushroom (Enok), a Drosophila putative MYST family histone acetyltransferase controls GSC maintenance in the ovary at multiple levels. Removal or knockdown of Enok in the germline causes a GSC maintenance defect. Further studies show that the cell-autonomous role of Enok in maintaining GSCs is not dependent on the BMP/Bam pathway. Interestingly, molecular studies reveal an ectopic expression of Bruno, an RNA binding protein, in the GSCs and their differentiating daughter cells elicited by the germline Enok deficiency. Misexpression of Bruno in GSCs and their immediate descendants results in a GSC loss that can be exacerbated by incorporating one copy of enok mutant allele. These data suggest a role for Bruno in Enok-controlled GSC maintenance. In addition, we observe that Enok is required for maintaining GSCs non-autonomously. Compromised expression of enok in the niche cells impairs the niche maintenance and BMP signal output, thereby causing defective GSC maintenance. This is the first demonstration that the niche size control requires an epigenetic mechanism. Taken together, studies in this paper provide new insights into the GSC fate regulation.
Collapse
Affiliation(s)
- Tianchi Xin
- MoE Key Laboratory of Developmental Genetics and Neuropsychiatric Diseases, Bio-X Institutes, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 200240 Shanghai, PR China
| | | | | | | | | | | |
Collapse
|
27
|
Dourlen P, Levet C, Mejat A, Gambis A, Mollereau B. The Tomato/GFP-FLP/FRT method for live imaging of mosaic adult Drosophila photoreceptor cells. J Vis Exp 2013:e50610. [PMID: 24084155 PMCID: PMC3923918 DOI: 10.3791/50610] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The Drosophila eye is widely used as a model for studies of development and neuronal degeneration. With the powerful mitotic recombination technique, elegant genetic screens based on clonal analysis have led to the identification of signaling pathways involved in eye development and photoreceptor (PR) differentiation at larval stages. We describe here the Tomato/GFP-FLP/FRT method, which can be used for rapid clonal analysis in the eye of living adult Drosophila. Fluorescent photoreceptor cells are imaged with the cornea neutralization technique, on retinas with mosaic clones generated by flipase-mediated recombination. This method has several major advantages over classical histological sectioning of the retina: it can be used for high-throughput screening and has proved an effective method for identifying the factors regulating PR survival and function. It can be used for kinetic analyses of PR degeneration in the same living animal over several weeks, to demonstrate the requirement for specific genes for PR survival or function in the adult fly. This method is also useful for addressing cell autonomy issues in developmental mutants, such as those in which the establishment of planar cell polarity is affected.
Collapse
Affiliation(s)
- Pierre Dourlen
- Laboratory of Molecular Biology of the Cell, Ecole Normale Supérieure de Lyon
| | | | | | | | | |
Collapse
|
28
|
Wnk kinases are positive regulators of canonical Wnt/β-catenin signalling. EMBO Rep 2013; 14:718-25. [PMID: 23797875 DOI: 10.1038/embor.2013.88] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 05/29/2013] [Accepted: 05/31/2013] [Indexed: 11/08/2022] Open
Abstract
Wnt/β-catenin signalling is central to development and its regulation is essential in preventing cancer. Using phosphorylation of Dishevelled as readout of pathway activation, we identified Drosophila Wnk kinase as a new regulator of canonical Wnt/β-catenin signalling. WNK kinases are known for regulating ion co-transporters associated with hypertension disorders. We demonstrate that wnk loss-of-function phenotypes resemble canonical Wnt pathway mutants, while Wnk overexpression causes gain-of-function canonical Wnt-signalling phenotypes. Importantly, knockdown of human WNK1 and WNK2 also results in decreased Wnt signalling in mammalian cell culture, suggesting that Wnk kinases have a conserved function in ensuring peak levels of canonical Wnt signalling.
Collapse
|
29
|
Steimel A, Suh J, Hussainkhel A, Deheshi S, Grants JM, Zapf R, Moerman DG, Taubert S, Hutter H. The C. elegans CDK8 Mediator module regulates axon guidance decisions in the ventral nerve cord and during dorsal axon navigation. Dev Biol 2013; 377:385-98. [PMID: 23458898 DOI: 10.1016/j.ydbio.2013.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 01/21/2013] [Accepted: 02/14/2013] [Indexed: 11/30/2022]
Abstract
Receptors expressed on the growth cone of outgrowing axons detect cues required for proper navigation. The pathway choices available to an axon are in part defined by the set of guidance receptors present on the growth cone. Regulated expression of receptors and genes controlling the localization and activity of receptors ensures that axons respond only to guidance cues relevant for reaching their targets. In genetic screens for axon guidance mutants, we isolated an allele of let-19/mdt-13, a component of the Mediator, a large ~30 subunit protein complex essential for gene transcription by RNA polymerase II. LET-19/MDT-13 is part of the CDK8 module of the Mediator. By testing other Mediator components, we found that all subunits of the CDK8 module as well as some other Mediator components are required for specific axon navigation decisions in a subset of neurons. Expression profiling demonstrated that let-19/mdt-13 regulates the expression of a large number of genes in interneurons. A mutation in the sax-3 gene, encoding a receptor for the repulsive guidance cue SLT-1, suppresses the commissure navigation defects found in cdk-8 mutants. This suggests that the CDK8 module specifically represses the SAX-3/ROBO pathway to ensure proper commissure navigation.
Collapse
Affiliation(s)
- Andreas Steimel
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Sato A, Shibuya H. WNK signaling is involved in neural development via Lhx8/Awh expression. PLoS One 2013; 8:e55301. [PMID: 23383144 PMCID: PMC3559379 DOI: 10.1371/journal.pone.0055301] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 12/20/2012] [Indexed: 02/07/2023] Open
Abstract
WNK kinase family is conserved among many species and regulates SPAK/OSR1 and ion co-transporters. Some mutations in human WNK1 or WNK4 are associated with Pseudohypoaldosteronism type II, a form of hypertension. WNK is also involved in developmental and cellular processes, but the molecular mechanisms underlying its regulation in these processes remain unknown. Here, we identify a new target gene in WNK signaling, Arrowhead and Lhx8, which is a mammalian homologue of Drosophila Arrowhead. In Drosophila, WNK was shown to genetically interact with Arrowhead. In Wnk1 knockout mice, levels of Lhx8 expression were reduced. Ectopic expression of WNK1, WNK4 or Osr1 in mammalian cells induced the expression of the Lhx8. Moreover, neural specification was inhibited by the knockdown of both Wnk1 and Wnk4 or Lhx8. Drosophila WNK mutant caused defects in axon guidance during embryogenesis. These results suggest that WNK signaling is involved in the morphological and neural development via Lhx8/Arrowhead.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of Molecular Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Shibuya
- Department of Molecular Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
31
|
St Johnston D. Using mutants, knockdowns, and transgenesis to investigate gene function in Drosophila. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:587-613. [PMID: 24014449 DOI: 10.1002/wdev.101] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The sophisticated genetic techniques available in Drosophila are largely responsible for its success as a model organism. One of the most important of these is the ability to disrupt gene function in vivo and observe the resulting phenotypes. This review considers the ever-increasing repertoire of approaches for perturbing the functions of specific genes in flies, ranging from classical and transposon-mediated mutageneses to newer techniques, such as homologous recombination and RNA interference. Since most genes are used over and over again in different contexts during development, many important advances have depended on being able to interfere with gene function at specific times or places in the developing animal, and a variety of approaches are now available to do this. Most of these techniques rely on being able to create genetically modified strains of Drosophila and the different methods for generating lines carrying single copy transgenic constructs will be described, along with the advantages and disadvantages of each approach.
Collapse
Affiliation(s)
- Daniel St Johnston
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Cambridge CB2 1QN, UK.
| |
Collapse
|
32
|
Nfonsam LE, Cano C, Mudge J, Schilkey FD, Curtiss J. Analysis of the transcriptomes downstream of Eyeless and the Hedgehog, Decapentaplegic and Notch signaling pathways in Drosophila melanogaster. PLoS One 2012; 7:e44583. [PMID: 22952997 PMCID: PMC3432130 DOI: 10.1371/journal.pone.0044583] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 08/09/2012] [Indexed: 01/22/2023] Open
Abstract
Tissue-specific transcription factors are thought to cooperate with signaling pathways to promote patterned tissue specification, in part by co-regulating transcription. The Drosophila melanogaster Pax6 homolog Eyeless forms a complex, incompletely understood regulatory network with the Hedgehog, Decapentaplegic and Notch signaling pathways to control eye-specific gene expression. We report a combinatorial approach, including mRNAseq and microarray analyses, to identify targets co-regulated by Eyeless and Hedgehog, Decapentaplegic or Notch. Multiple analyses suggest that the transcriptomes resulting from co-misexpression of Eyeless+signaling factors provide a more complete picture of eye development compared to previous efforts involving Eyeless alone: (1) Principal components analysis and two-way hierarchical clustering revealed that the Eyeless+signaling factor transcriptomes are closer to the eye control transcriptome than when Eyeless is misexpressed alone; (2) more genes are upregulated at least three-fold in response to Eyeless+signaling factors compared to Eyeless alone; (3) based on gene ontology analysis, the genes upregulated in response to Eyeless+signaling factors had a greater diversity of functions compared to Eyeless alone. Through a secondary screen that utilized RNA interference, we show that the predicted gene CG4721 has a role in eye development. CG4721 encodes a neprilysin family metalloprotease that is highly up-regulated in response to Eyeless+Notch, confirming the validity of our approach. Given the similarity between D. melanogaster and vertebrate eye development, the large number of novel genes identified as potential targets of Ey+signaling factors will provide novel insights to our understanding of eye development in D. melanogaster and humans.
Collapse
Affiliation(s)
- Landry E. Nfonsam
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Carlos Cano
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Joann Mudge
- National Center for Genome Resources, Santa Fe, New Mexico, United States of America
| | - Faye D. Schilkey
- National Center for Genome Resources, Santa Fe, New Mexico, United States of America
| | - Jennifer Curtiss
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| |
Collapse
|
33
|
Abstract
Both insect and vertebrate visual circuits are organized into orderly arrays of columnar and layered synaptic units that correspond to the array of photoreceptors in the eye. Recent genetic studies in Drosophila have yielded insights into the molecular and cellular mechanisms that pattern the layers and columns and establish specific connections within the synaptic units. A sequence of inductive events and complex cellular interactions coordinates the assembly of visual circuits. Photoreceptor-derived ligands, such as hedgehog and Jelly-Belly, induce target development and expression of specific adhesion molecules, which in turn serve as guidance cues for photoreceptor axons. Afferents are directed to specific layers by adhesive afferent-target interactions mediated by leucine-rich repeat proteins and cadherins, which are restricted spatially and/or modulated dynamically. Afferents are restricted to their topographically appropriate columns by repulsive interactions between afferents and by autocrine activin signaling. Finally, Dscam-mediated repulsive interactions between target neuron dendrites ensure appropriate combinations of postsynaptic elements at synapses. Essentially, all these Drosophila molecules have vertebrate homologs, some of which are known to carry out analogous functions. Thus, the studies of Drosophila visual circuit development would shed light on neural circuit assembly in general.
Collapse
Affiliation(s)
- Krishna V Melnattur
- Section on Neuronal Connectivity, Laboratory of Gene Regulation and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
34
|
Distefano GM, Gangemi AJ, Khandelwal PJ, Saunders AJ, Marenda DR. Drosophila lilliputian is required for proneural gene expression in retinal development. Dev Dyn 2012; 241:553-62. [PMID: 22275119 DOI: 10.1002/dvdy.23738] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2011] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Proper neurogenesis in the developing Drosophila retina requires the regulated expression of the basic helix-loop-helix (bHLH) proneural transcription factors Atonal (Ato) and Daughterless (Da). Factors that control the timing and spatial expression of these bHLH proneural genes in the retina are required for the proper formation and function of the adult eye and nervous system. RESULTS Here we report that lilliputian (lilli), the Drosophila homolog of the FMR2/AF4 family of proteins, regulates the transcription of ato and da in the developing fly retina. We find that lilli controls ato expression at multiple enhancer elements. We also find that lilli contributes to ato auto-regulation in the morphogenetic furrow by first regulating the expression of da prior to ato. We show that FMR2 regulates the ato and da homologs MATH5 and TCF12 in human cells, suggesting a conservation of this regulation from flies to humans. CONCLUSIONS We conclude that lilliputian is part of the genetic program that regulates the expression of proneural genes in the developing retina.
Collapse
Affiliation(s)
- Ginnene M Distefano
- Department of Biology, Drexel University, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
35
|
Gontang AC, Hwa JJ, Mast JD, Schwabe T, Clandinin TR. The cytoskeletal regulator Genghis khan is required for columnar target specificity in the Drosophila visual system. Development 2011; 138:4899-909. [PMID: 22007130 DOI: 10.1242/dev.069930] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A defining characteristic of neuronal cell type is the growth of axons and dendrites into specific layers and columns of the brain. Although differences in cell surface receptors and adhesion molecules are known to cause differences in synaptic specificity, differences in downstream signaling mechanisms that determine cell type-appropriate targeting patterns are unknown. Using a forward genetic screen in Drosophila, we identify the GTPase effector Genghis khan (Gek) as playing a crucial role in the ability of a subset of photoreceptor (R cell) axons to innervate appropriate target columns. In particular, single-cell mosaic analyses demonstrate that R cell growth cones lacking Gek function grow to the appropriate ganglion, but frequently fail to innervate the correct target column. Further studies reveal that R cell axons lacking the activity of the small GTPase Cdc42 display similar defects, providing evidence that these proteins regulate a common set of processes. Gek is expressed in all R cells, and a detailed structure-function analysis reveals a set of regulatory domains with activities that restrict Gek function to the growth cone. Although Gek does not normally regulate layer-specific targeting, ectopic expression of Gek is sufficient to alter the targeting choices made by another R cell type, the targeting of which is normally Gek independent. Thus, specific regulation of cytoskeletal responses to targeting cues is necessary for cell type-appropriate synaptic specificity.
Collapse
Affiliation(s)
- Allison C Gontang
- Department of Neurobiology, 299 W. Campus Drive, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
36
|
Stephan R, Gohl C, Fleige A, Klämbt C, Bogdan S. Membrane-targeted WAVE mediates photoreceptor axon targeting in the absence of the WAVE complex in Drosophila. Mol Biol Cell 2011; 22:4079-92. [PMID: 21900504 PMCID: PMC3204070 DOI: 10.1091/mbc.e11-02-0121] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A tight spatial-temporal coordination of F-actin dynamics is crucial for a large variety of cellular processes that shape cells. The Abelson interactor (Abi) has a conserved role in Arp2/3-dependent actin polymerization, regulating Wiskott-Aldrich syndrome protein (WASP) and WASP family verprolin-homologous protein (WAVE). In this paper, we report that Abi exerts nonautonomous control of photoreceptor axon targeting in the Drosophila visual system through WAVE. In abi mutants, WAVE is unstable but restored by reexpression of Abi, confirming that Abi controls the integrity of the WAVE complex in vivo. Remarkably, expression of a membrane-tethered WAVE protein rescues the axonal projection defects of abi mutants in the absence of the other subunits of the WAVE complex, whereas cytoplasmic WAVE only slightly affects the abi mutant phenotype. Thus complex formation not only stabilizes WAVE, but also provides further membrane-recruiting signals, resulting in an activation of WAVE.
Collapse
Affiliation(s)
- Raiko Stephan
- Institut für Neurobiologie, Universität Münster, 48149 Münster, Germany
| | | | | | | | | |
Collapse
|
37
|
Ohler S, Hakeda-Suzuki S, Suzuki T. Hts, the Drosophila homologue of Adducin, physically interacts with the transmembrane receptor Golden goal to guide photoreceptor axons. Dev Dyn 2011; 240:135-48. [PMID: 21128303 DOI: 10.1002/dvdy.22515] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Neurons steer their axons towards their proper targets during development. Molecularly, a number of guidance receptors have been identified. The transmembrane protein Golden goal (Gogo) was reported previously to guide photoreceptor (R) axons in the Drosophila visual system. Here, we show that Hts, the Drosophila homologue of Adducin, physically interacts with Gogo's cytoplasmic domain via its head-neck domain. hts null mutants show similar defects in R axon guidance as do gogo mutants. Rescue experiments suggest that the C-terminal tail but not the MARCKS homology domain of Hts is required. Overexpression of either gogo or hts causes abnormally thick swellings of R8 axons in the medulla, but if both are co-overexpressed, R8 axons appear normal and the amount of excessive Hts is reduced. Our results fit with a model where Gogo both positively and negatively regulates Hts that affects the Actin-Spectrin cytoskeleton in growth cone filopodia, thereby guiding R axons.
Collapse
Affiliation(s)
- Stephan Ohler
- Max Planck Institute of Neurobiology, Martinsried, Germany
| | | | | |
Collapse
|
38
|
Golden Goal collaborates with Flamingo in conferring synaptic-layer specificity in the visual system. Nat Neurosci 2011; 14:314-23. [PMID: 21317905 DOI: 10.1038/nn.2756] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 01/14/2011] [Indexed: 01/06/2023]
Abstract
Neuronal connections are often organized in layers that contain synapses between neurons that have similar functions. In Drosophila, R7 and R8 photoreceptors, which detect different wavelengths, form synapses in distinct medulla layers. The mechanisms underlying the specificity of synaptic-layer selection remain unclear. We found that Golden Goal (Gogo) and Flamingo (Fmi), two cell-surface proteins involved in photoreceptor targeting, functionally interact in R8 photoreceptor axons. Our results indicate that Gogo promotes R8 photoreceptor axon adhesion to the temporary layer M1, whereas Gogo and Fmi collaborate to mediate axon targeting to the final layer M3. Structure-function analysis suggested that Gogo and Fmi interact with intracellular components through the Gogo cytoplasmic domain. Moreover, Fmi was also required in target cells for R8 photoreceptor axon targeting. We propose that Gogo acts as a functional partner of Fmi for R8 photoreceptor axon targeting and that the dynamic regulation of their interaction specifies synaptic-layer selection of photoreceptors.
Collapse
|
39
|
Hadjieconomou D, Timofeev K, Salecker I. A step-by-step guide to visual circuit assembly in Drosophila. Curr Opin Neurobiol 2011; 21:76-84. [PMID: 20800474 DOI: 10.1016/j.conb.2010.07.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 07/27/2010] [Indexed: 01/03/2023]
Affiliation(s)
- Dafni Hadjieconomou
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, The Ridgeway, NW7 1AA London, United Kingdom
| | | | | |
Collapse
|
40
|
CDK19 is disrupted in a female patient with bilateral congenital retinal folds, microcephaly and mild mental retardation. Hum Genet 2010; 128:281-91. [PMID: 20563892 PMCID: PMC2921488 DOI: 10.1007/s00439-010-0848-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 06/03/2010] [Indexed: 12/13/2022]
Abstract
Microcephaly, mental retardation and congenital retinal folds along with other systemic features have previously been reported as a separate clinical entity. The sporadic nature of the syndrome and lack of clear inheritance patterns pointed to a genetic heterogeneity. Here, we report a genetic analysis of a female patient with microcephaly, congenital bilateral falciform retinal folds, nystagmus, and mental retardation. Karyotyping revealed a de novo pericentric inversion in chromosome 6 with breakpoints in 6p12.1 and 6q21. Fluorescence in situ hybridization analysis narrowed down the region around the breakpoints, and the breakpoint at 6q21 was found to disrupt the CDK19 gene. CDK19 was found to be expressed in a diverse range of tissues including fetal eye and fetal brain. Quantitative PCR of the CDK19 transcript from Epstein–Barr virus-transformed lymphoblastoid cell lines of the patient revealed ~50% reduction in the transcript (p = 0.02), suggesting haploinsufficiency of the gene. cdk8, the closest orthologue of human CDK19 in Drosophila has been shown to play a major role in eye development. Conditional knock-down of Drosophila cdk8 in multiple dendrite (md) neurons resulted in 35% reduced dendritic branching and altered morphology of the dendritic arbour, which appeared to be due in part to a loss of small higher order branches. In addition, Cdk8 mutant md neurons showed diminished dendritic fields revealing an important role of the CDK19 orthologue in the developing nervous system of Drosophila. This is the first time the CDK19 gene, a component of the mediator co-activator complex, has been linked to a human disease.
Collapse
|
41
|
Regulation of axonal development by the nuclear protein hindsight (pebbled) in the Drosophila visual system. Dev Biol 2010; 344:911-21. [PMID: 20541542 DOI: 10.1016/j.ydbio.2010.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 05/14/2010] [Accepted: 06/03/2010] [Indexed: 11/24/2022]
Abstract
The molecules and networks involved in the process of acquisition and maintenance of the form of a mature neuron are not completely known. Using a misexpression screen we identified the gene hindsight as a gene involved in the process of acquisition of the neuronal morphogenesis in the Drosophila adult nervous system. hindsight encodes a transcription factor known for its role in early developmental processes such as embryonic germ band retraction and dorsal closure, as well as in the establishment of cell morphology, planar cell polarity, and epithelial integrity during retinal development. We describe here a novel function for HNT by showing that both loss and gain of function of HNT affects the pathfinding of the photoreceptors axons. By manipulating the timing and level of HNT expression, together with the number of cells manipulated we show here that the function of HNT in axonal guidance is independent of the HNT functions previously reported in retinal cells. Based on genetic interaction experiments we show that part of HNT function in axonal development is exerted through the regulation of genes involved in the dynamics of the actin cytoskeleton.
Collapse
|
42
|
Astigarraga S, Hofmeyer K, Treisman JE. Missed connections: photoreceptor axon seeks target neuron for synaptogenesis. Curr Opin Genet Dev 2010; 20:400-7. [PMID: 20434326 DOI: 10.1016/j.gde.2010.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2010] [Revised: 03/31/2010] [Accepted: 04/02/2010] [Indexed: 02/06/2023]
Abstract
Extending axons must choose the appropriate synaptic target cells in order to assemble functional neural circuitry. The axons of the Drosophila color-sensitive photoreceptors R7 and R8 project as a single fascicle from each ommatidium, but their terminals are segregated into distinct layers within their target region. Recent studies have begun to reveal the molecular mechanisms that establish this projection pattern. Both homophilic adhesion molecules and specific ligand-receptor interactions make important contributions to stabilizing R7 and R8 terminals in the appropriate target layers. These cell recognition molecules are regulated by the same transcription factors that control R7 and R8 cell fates. Autocrine and repulsive signaling mechanisms prevent photoreceptor terminals from encroaching on their neighbors, preserving the spatial resolution of visual information.
Collapse
Affiliation(s)
- Sergio Astigarraga
- Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | | | | |
Collapse
|
43
|
Rallis A, Moore C, Ng J. Signal strength and signal duration define two distinct aspects of JNK-regulated axon stability. Dev Biol 2009; 339:65-77. [PMID: 20035736 PMCID: PMC2845820 DOI: 10.1016/j.ydbio.2009.12.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 12/10/2009] [Accepted: 12/11/2009] [Indexed: 11/29/2022]
Abstract
Signaling proteins often control multiple aspects of cell morphogenesis. Yet the mechanisms that govern their pleiotropic behavior are often unclear. Here we show activity levels and timing mechanisms determine distinct aspects of Jun N-terminal kinase (JNK) pathway dependent axonal morphogenesis in Drosophila mushroom body (MB) neurons. In the complete absence of Drosophila JNK (Basket), MB axons fail to stabilize, leading to their subsequent degeneration. However, with a partial loss of Basket (Bsk), or of one of the upstream JNK kinases, Hemipterous or Mkk4, these axons overextend. This suggests that Bsk activity prevents axons from destabilizing, resulting in degeneration and overextension beyond their terminal targets. These distinct phenotypes require different threshold activities involving the convergent action of two distinct JNK kinases. We show that sustained Bsk signals are essential throughout development and act additively but are dispensable at adulthood. We also suggest that graded Bsk inputs are translated into AP-1 transcriptional outputs consisting of Fos and Jun proteins.
Collapse
Affiliation(s)
- Andrew Rallis
- MRC Centre for Developmental Neurobiology, New Hunt's House, Guy's Campus, King's College, London SE1 1UL, UK
| | | | | |
Collapse
|